You are on page 1of 10

Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Ginger and its active compounds in cancer therapy: From folk uses to nano-
therapeutic applications
M.F. Mahomoodallya, M.Z. Aumeeruddya, Kannan R.R. Rengasamyb,⁎, S. Roshanc, S. Hammadd,e,
J. Pandoheea,f, Xuebo Hug, G. Zenginh
a
Department of Health Sciences, Faculty of Science, University of Mauritius, 230 Réduit, Mauritius
b
Department of Bioresources and Food Science, College of Life Sciences, Konkuk University, Seoul 05029, South Korea
c
Deccan School of Pharmacy, Darussalam, Aghapura, Hyderabad, 500001, Telangana, India
d
School of Pharmacy, Monash University, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia
e
Institute of Pharmaceutical Sciences (IPS), University of Veterinary & Animal Sciences (UVAS), Lahore, Pakistan
f
Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Joondalup, WA 6027, Australia
g
College of Plant Sciences and Technology, Huazhong Agricultural University, Wuhan, China
h
Department of Biology, Faculty of Science, Selcuk University, Turkey

ARTICLE INFO ABSTRACT

Keywords: Ginger is a spice that is renowned for its characteristic aromatic fragrance and pungent taste, with documented
Zingiber officinale healing properties. Field studies conducted in several Asian and African countries revealed that ginger is used
Microbiome traditionally in the management of cancer. The scientific community has probed into the biological validation of
Gingerol its extracts and isolated compounds including the gingerols, shogaols, zingiberene, and zingerone, through in-
Shogaol
vitro and in-vivo studies. Nonetheless, an updated compilation of these data together with a deep mechanistic
Enzyme inhibition
Combination therapy
approach is yet to be provided. Accordingly, this review highlights the mechanisms and therapeutics of ginger
Nanoformulation and its bioactive compounds focused on a cancer context and these evidence are based on the (i) cytotoxic effect
against cancer cell lines, (ii) enzyme inhibitory action, (iii) combination therapy with chemotherapeutic and
phenolic compounds, (iv) possible links to the microbiome and (v) the use of nano-formulations of ginger
bioactive compounds as a more effective drug delivery strategy in cancer therapy.

1. Introduction carbohydrates, water, protein, lipids, fibres and volatile essential oils
[2,3]. Using advanced mass spectrometry-based identifications, che-
The ginger plant is a herbaceous flowering plant that belongs to the mical analysis of the essential oil reveals that the root contains hun-
Zingiberaceae family. Scientifically named as Zingiber officinale Roscoe, dreds of different compounds [4,5]. The majority of compounds iden-
the perennial plant consists of a pseudo-stem, yellow flowers and tu- tified in ginger are from the chemical classes gingerols, shogaols,
berous rhizomes, which are also known as ginger root or commonly zigiberenes and zingerone (chemical structures shown in Fig. 1). To a
called ginger. It is the rhizomes of ginger plants that are the most lesser extent, terpene components such as bisabolene, farnesene, ses-
sought-after portion due to its aromatic odour and pungent taste. quiphellandrene, curcumene and phytosterols, vitamins and minerals
Ginger is therefore an important ingredient for culinary purposes. can also be found in specific varieties of ginger. Thus, the nutraceutical
Originally from Asia, the most common custom usage of ginger was as a value of ginger is attributed to its complexity of bioactive compounds
flavouring agent in its various form and this could include fresh, dried, especially the major phenolic classes such as the gingerols, shogaols,
pickled, powdered and preserved and, more interestingly, as a tonic zingiberene, paradol, and zingerone [6].
root to treat many ailments for its medicinal benefits. Nowadays, the Gingerols (23–25%) are part of the phenolic compound class and
plant is grown on a large scale across the world in tropical regions, such these volatile organic compounds are known to account for the strong
as the West Indies, Africa and India, for its consumption as a spice, taste of fresh Zingiber officinale. 6-gingerol is the main compound ac-
dietary supplement [1]. countable for the pungency of the rhizome, while the other gingerols
As a natural product, ginger is a complex spice composed of (4-, 8-, 10- and 12-gingerol) are present in lower amounts (1–10%).


Corresponding author.
E-mail address: Rengasamy@enzymeinhibitors.co.in (K.R.R. Rengasamy).

https://doi.org/10.1016/j.semcancer.2019.08.009
Received 3 July 2019; Received in revised form 26 July 2019; Accepted 9 August 2019
1044-579X/ © 2019 Elsevier Ltd. All rights reserved.

Please cite this article as: M.F. Mahomoodally, et al., Seminars in Cancer Biology, https://doi.org/10.1016/j.semcancer.2019.08.009
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Fig. 1. The chemical structures of gingerol, shogal, gingerdione and zingerol.

Fig. 2. Cytotoxic mechanisms of action of two studied compounds in Ginger.

However, being thermally labile, these compounds are converted to Using ginger in the treatment of cancer is not a new concept, it has
shogaols at high temperatures, for example while cooking, and gives been a customary practice in the folk medicine throughout the world
ginger its spicy-sweet aroma. The biological properties of gingerols and and has been showed to be effective by several field studies. In India,
shogaols are recognized to possess antimicrobial, anticancer, anti- the juice and decoction of the rhizome is taken orally [10,11]. In Sin-
oxidant, anti-inflammatory, and anti-allergic propensities [7,8]. Sho- gapore, the cooked rhizome is used as a mean for cancer prevention
gaols are known to have anti-coughing effects, gingerol on the other [12]. An infusion of the rhizome is used against breast cancer by the
hand accounts for the analgesic properties of ginger. In addition to the Palestinians [13]. In addition, as a general cancer treatment, a decoc-
phenolics, diarylheptanoids and zingerone have also been identified in tion of the root is prepared from a mix of ginger root, turmeric, and
ginger and are believed to be bioactive compounds contributing to honey, and two doses are ingested on a daily basis. A concoction can
health benefits [5,6,9]. also be made from a mix of ginger root, nigella, and camel milk, and

2
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

one glass is taken daily before breakfast [14]. Another recipe used by cytotoxic than 6-shogaol (IC50 = 100.0 μM) against PC-3 cell [22]. In
the Palestinians to manage stomach and liver cancer uses 100 g of the addition, at a concentration of 40 μmol/L, 6-shogaol abridged the sur-
ground dried rhizomes boiled in water and given twice daily after meals vival of DU145, LNCaP, and PC-3 by 80%, 96%, and 76%, respectively,
[13]. In Morocco, ginger root is ground with honey and taken orally after 72 h exposure [24]. The authors also found that 6-shogaol in-
[15]. Another traditional use of ginger is as a paste; in Ghana the root/ hibited STAT3 and NF-kB signaling, decreased interleukin (IL)-6-in-
rhizome is ground and the resulting mixture is either orally consumed duced STAT3 initiation and also inhibited TNF-α-induced NF-kB ac-
or used as a massage remedy for stomach and brain cancer [16]. tivity. Moreover, 6-shogaol decreased the level of numerous STAT3 and
Recently, pharmacological studies have probed into the validation NF-kB-regulated target genes at the proteinomic stage, such as cyclin
of such claims, and now, ginger together with its associated bioactive D1, survivin, and cMyc. 6-shogaol also modulated the mRNA levels of
compounds have been proven to have potential anticancer properties. several chemokine, cytokine, cell cycle, and apoptosis regulatory genes
In this context, this review aims at highlighting the cytotoxic effect of including IL-7, CCL5, BAX, BCL2, p21, and p27. 6-gingerol also induced
ginger and its compounds against various types of cancer, focusing on dose- and time-dependent apoptosis in LNCaP human prostate cancer
its enzyme inhibitory mechanism, combination therapy with other cells mainly via caspase-3 expression with the resulting degradation of
agents and the potential of nano-formulations of ginger bioactive PARP [25].
compounds as a novel beneficial in cancer treatment.
2.4. Lung cancer
2. Overview of anti-cancer properties of ginger
The cytotoxic effect of 6-gingerol (IC50 = 136.73 μM after 24 h ex-
There has been an increasing amount of studies showing promising
posure) on H-1299 lung cancer cell was proved by Lv et al [26]. Peng
results on the cytotoxic effect of ginger extracts and its bioactive
et al [22] found that 6-shogaol (IC50 = 22.9 μM) was more effective
compounds against several cancer cells including breast, cervical, col-
than 10-gingerol (IC50 = 85.4 μM) against A549 lung cancer cell. When
orectal, leukemia, liver, lung, nasopharyngeal, ovarian, prostate, and
evaluated against human lung cancer cells, 6-shogaol was also observed
retinoblastoma (see detailed findings and mechanisms in Table 1 and
to initiate autophagy via the inhibition of AKT/mTOR pathway [27].
Fig. 2).

2.1. Breast cancer 2.5. Liver cancer

Ansari et al [17] showed that the methanolic solution of ginger Ginger extract was observed to diminish the raised activity of NFκB
displayed a time-dependent cytotoxic effect on the breast cancer cell and TNF-α in rats with liver cancer [28]. In addition, other ginger
line (MDA-MB-231), with IC50 of 86.7 and 57.5 μg/mL after 24 and 48 h derivatives such as Z-6-oxo-6-shogaol, Z-6-oxo-8-shogaol and E-4-iso-
incubation, respectively. In addition, the two derivatives of 6-oxo- shogaol, exhibited cytotoxic effect against HepG2 cell, with IC50 scores
shogaol isolated from ginger, namely Z-6-oxo-6-shogaol and Z-6-oxo-8- of 8.92 μM, 45.14 μM, 14.87 μM, respectively [5], which was more
shogaol, showed potent cytotoxic action on MCF-7 cell, with IC50 of effective than the essential oil (IC50 = 635.1 μg/mL) [29]. 72 h ex-
6.27 and 47.22 μM, respectively, after 48 h exposure [5]. 6-shogaol was posure of 6-shogaol also showed higher cytotoxic effect on BEL-7404
found to hinder the growth of cancer cell (breast & colon) by activating compared to 10-gingerol [22]·
the peroxisomal proliferator activated receptor γ (PPARγ) [18]. It has
been observed [19] that 10-gingerol caused a considerable upsurge in
2.6. Blood cancer
the initiation of caspase-3 and inhibited orthotopic tumour growth of
spontaneous breast cancer metastasis. More pertinently, the authors
6-shogaol was a more effective cytotoxic agent against HL-60 and
reported that 10-gingerol inhibit metastasis to multiple organs (in-
K562 leukemic cell (supported by IC50 values of 7.9 μM and 24.2 μM,
cluding lung, bone, and brain).
respectively), which was more effective compared to 10-gingerol [22].
Also, Liu et al [30] observed that 6-shogaol specifically stimulated
2.2. Cervical cancer
apoptosis in transformed and primary cellular leukemia. Data from
immunoblotting studies revealed that 6-shogaol caused apoptosis by a
A study by Liu et al [20] found that 6-shogaol exhibited high in-
process which involves eIF2α dephosphorylation and caspase activa-
hibition (IC50 = 14.75 μM) against HeLa cells, which was more effec-
tion-dependent breakdown of eIF2α.
tive compared to α-zingiberene (IC50 = 60.6 μg/mL), 6-gingerol
(IC50 = 96.32 μM), and [10]-gingerol (IC50 = 52.4 μM) [21–23]. The
essential oil was also found to be cytotoxic to SiHa cells, showing IC50 2.7. Colorectal cancer
value of 38.6 μg/mL, more effective than α-zingiberene
(IC50 = 46.2 μg/mL) [21]. 6-gingerol blocked the cell cycle in G0/G1- A plethora of supporting information that ginger and its bioactive
phase in HeLa cell and eventually caused cells death. The compound derivatives are cytotoxic to several colon cancer cell lines with HCT15,
reduced cyclin (A, D1, E1) expression, slightly decreased CDK-1, p21, HCT116, SW480, LoVo, and HT29 cells (see Table 1). It has been [31]
and p27 while cyclin B1 and E1 protein were unaffected. Western blot observed that 6-gingerol caused apoptosis in colorectal cancer cells by
analysis also indicated the pathway of apoptotic activation, with an upregulating NAG-1 and G1 cell cycle arrest via the down-regulation of
increased Bax/Bcl-2 ratio, cytochrome c release, cleavage of caspase-3, cyclin D1. Several mechanisms seem to be responsible for the action of
-8, -9 and phosphoribosyl pyrophosphate (PRPP) in challenged cells. 6-gingerol, including protein degradation along with β-catenin, PKCε,
Consequently, 6-gingerol triggered AMP-activated protein kinase and GSK-3β pathways. 6-gingerol also stimulated caspase-dependent
(AMPK), and inhibited PI3K/AKT phosphorylation, thus leading to a apoptosis and prevented phorbol myristate acetate-induced prolifera-
decrease in P70S6K expression and ultimately inhibited mTOR [23]. tion in colon cancer cells through the inhibition of MAPK/AP-1 sig-
naling. On top of that, using p53(-/-) and p53(+/+) HCT-116 color-
2.3. Prostate cancer ectal cancer cells, the researchers confirmed that p53/p21 was the
principal pathway contributing to the G2/M cell cycle arrest induced by
Ginger and its active compounds were also found to reduce the cell 6-shogaol [32]. It was [33] found that 6-shogaol induced-apoptosis
viability of a number of prostate cancer cells including DU145, LNCaP, primarily via the pathway involving the mitochondria, and the Bcl-2
C4-2, C4-2B, and PC-3. 10-gingerol (IC50 = 59.7 μM) was more family might play as a central regulator.

3
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 1
Cytotoxicity of ginger and bioactive compounds against cancer cell lines.
Cancer type Cell line Extract type/compound Incubation time Findings References

Breast MDA-MB-231 Methanol extract 24 h and 48 h 24 h: IC50 = 86.7 μg/mL [17]


48 h: IC50 = 57.5 μg/mL
Ethyl acetate fraction 24 h At 100 μg/mL, reduced the cell viability by 44% [43]
obtained from crude
methanolic extract
MCF-7 Ethyl acetate fraction 24 h At 100 μg/mL, reduced the cell viability by 36% [43]
obtained from crude
methanolic extract
Essential oil 24 h IC50 = 82.6 μg/mL [21]
(Z)-6-oxo- [6]-shogaol 48 h IC50 = 6.27 μM [5]
(Z)-6-oxo- [8]-shogaol 48 h IC50 = 47.22 μM [5]
α-zingiberene 24 h IC50 = 172.0 μg/mL [21]
Cervical HeLa Chloroform extract 48 h IC50 = 74.32 μg/mL [44]
Ethanol extract 48 h IC50 = 33.78 μg/mL [44]
Essential oil 24 h MTT assay; IC50 = 141.4 μg/mL [29]
NRU assay; IC50 = 129.9 μg/mL
24 h IC50 = 46.0 μg/mL [21]
Methanol extract 24 h and 48 h 24 h: IC50 = 46.5 μg/mL [17]
48 h: IC50 = 37.5 μg/mL
6-shogaol 24 h IC50 = 14.75 μM [20]
72 h IC50 = 62.5 μM [22]
α-zingiberene 24 h IC50 = 60.6 μg/mL [21]
6-gingerol 48 h IC50 = 96.32 μM [23]
[10]-gingerol 72 h IC50 = 52.4 μM [22]
SiHa α-zingiberene 24 h IC50 = 46.2 μg/mL [21]
Essential oil 24 h IC50 = 38.6 μg/mL
Colorectal HCT15 6-Gingerol 24 h IC50 = 100 μM [45]
HCT-116 6-Gingerol 24 h IC50 = 160.42 μM [26]
Ethyl acetate fraction 24 h and 48 h Reduced the viability by 79 % and 88% at 200 μg/mL at 24 h and [43]
obtained from crude 48 h, respectively
methanolic extract
Ethanol extract 24 h IC50 = 496 μg/mL [46]
[6]-Gingerol 48 h,72 h or 96 Dose- and and time-dependent cytotoxic effect, with an IC50 value of [32]
h 283 μM
SW480 Ethyl acetate fraction 24 h and 48 h Reduced the viability by 55 % and 76% at 200 μg/mL at 24 h and [43]
obtained from crude 48 h, respectively
methanolic extract
[10]-Gingerol 24 h With 50-100 μM, [10]-gingerol reduced the cell viability in a [47]
concentration-dependent manner
[6]-Gingerol 48 h,72 h or 96 Dose- and and time-dependent cytotoxic effect, with an IC50 value of [32]
h 205 μM
LoVo Ethyl acetate fraction 24 h and 48 h Reduced the viability by 59 % and 80% at 200 μg/mL at 24 h and [43]
obtained from crude 48 h, respectively
methanolic extract
HT 29 Ethanol extract 24 h IC50 = 455 μg/mL [46]
Mucus-secreting Essential oil 24 h IC50 = 40 μL/mL [48]
HT29
Non-mucus- Essential oil 24 h IC50 = 60 μL/mL [48]
secreting HT-29
Leukemia Raw 264.7 cell 6-Gingerol 24 h IC50 = 102 μM [45]
HL-60 α-zingiberene 24 h IC50 = 80.3 μg/mL [21]
Essential oil 24 h IC50 = 39.1 μg/mL [21]
6-shogaol 72 h IC50 = 7.9 μM [22]
[10]-gingerol 72 h IC50 = 75.4 μM [22]
K562 6-shogaol 72 h IC50 = 24.2 μM [22]
[10]-gingerol 72 h IC50 = 112.5 μM [22]
Liver HepG2 Essential oil 24 h IC50 = 635.1 μg/mL [29]
Ethyl acetate fraction 24 h At 100 μg/mL, reduced the cell viability by 30% [43]
obtained from crude
methanolic extract
(Z)-6-oxo- [6]-shogaol 48 h IC50 = 8.92 μM [5]
(Z)-6-oxo- [8]-shogaol 48 h IC50 = 45.14 μM [5]
(E)- [4]-isoshogaol 48 h IC50 = 14.87 μM [5]
BEL-7404 6-shogaol 72 h IC50 = 11.8 μM [22]
[10]-gingerol 72 h IC50 = 95.2 μM [22]
Lung H-1299 6-gingerol 24 h IC50 = 136.73 μM [26]
A549 6-shogaol 72 h IC50 = 22.9 μM [22]
[10]-gingerol 72 h IC50 = 85.4 μM [22]
Murine fibrosarcoma L929 Essential oil 3h IC50 = 41 μg/mL [49]
Chloroform extract 48 h IC50 = 87.28 μg/mL [44]
Ethanol extract 48 h IC50 = 101.0 μg/mL [44]
6-Gingerol 24 h IC50 = 102 μM [45]
Nasopharyngeal CNE 6-shogaol 72 h IC50 = 43.8 μM [22]
[10]-gingerol 72 h IC50 = 88.1 μM [22]
(continued on next page)

4
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 1 (continued)

Cancer type Cell line Extract type/compound Incubation time Findings References

Ovarian SKOV-3 Extract (Sigma-Aldrich: 24 h, 48 h and IC50 was 97 μg/mL, 60 μg/mL and 40 μg/mL at 24 h, 48 h and 72 h, [50]
W252108) 72 h respectively
Prostate DU145 Methanolic fraction 24 h IC50 = 45.3 μg/mL [51]
Methanol extract 72 h IC50 = 95 μg/mL [52]
6-Shogaol 24 h, 48 h, and Reduced in % survival of cells, at a concentration of 40 mmol/L, was [24]
72 h 64%, 80%, and 80% after 24 h, 48 h, and 72 h, respectively
LNCaP Methanol extract 72 h IC50 = 75 μg/mL [52]
6-Shogaol 24 h, 48 h, and Reduced in % survival of cells, at a concentration of 40 mmol/L, was [24]
72 h 67%, 85%, and 96% after 24 h, 48 h, and 72 h, respectively
C4-2 Methanol extract 72 h IC50 = 512 μg/mL [52]
C4-2B Methanol extract 72 h IC50 = 240 μg/mL [52]
PC-3 Methanol extract 72 h IC50 = 250 μg/mL [52]
6-Shogaol 24 h, 48 h, and Reduced in % survival of cells, at a concentration of 40 mmol/L, was [24]
72 h 66%, 78%, and 76% after 24 h, 48 h, and 72 h, respectively
72 h IC50 = 100.0 μM [22]
[10]-gingerol 72 h IC50 = 59.7 μM [22]
Retinoblastoma RB355 Gingerol 12 h, 48 h, and Showed a concentration-dependent as well as time-dependent growth [53]
72 h inhibition. A 250-μM dose of gingerol led to 82.7%, 83.1%, and 85.2%
growth inhibition at 12-h, 48-h, and 72-h incubations

3. Links to the microbiome damage, which is the cause of many cancers, and especially in its po-
tential to control signaling molecules such as NF-κB, STAT3, MAPK,
With the increasing evidence that ginger has beneficial effects on PI3K, ERK1/2, Akt, TNF-α, COX-2, cyclin D1, cdk, MMP-9, surviving,
colorectal and intestinal cancer, the search to demystify the links and cIAP-1, XIAP and Bcl-2.
relationship between the bioactive compounds present in ginger and
the microbiome is becoming more and more critical. The microbiome is 4. Enzyme inhibition
the set of microorganisms that live inside a system; the human body
itself is home to 10–100 trillion symbiotic microbial cells, most of A number of enzymes are presently targeted as biomarkers in the
which live in the human gut alone [34]. It estimated that the total diagnosis or treatment of cancer. Targeting cellular metabolism asso-
amount of bacterial cells living in a human significantly outnumber ciated enzymes linked to cancer progression is considered to offer much
human cells and that microbes in the human intestines play a vital role promise but remains a largely unexplored topic [54]·
in maintaining a healthy gut function. Some of these functional con-
tributions include harvesting essential nutrients from non-digestible 4.1. Cytochrome P450
food such as fibres [35], vitamin synthesis [36] and the immune system
[37]. The core human microbiome is in constant interaction with its Cytochrome 450, shortened to CYP450, comprises a collection of
host and is susceptible to changes in various factors such as the host enzymes whose main function is the oxidative catalysis of numerous
lifestyle, diet, genotype, physiology, environment and disease status. In endogenous and exogenous constituents. Such enzymes are involved in
other words, the microbiome and its human host are in a symbiotic the phase I metabolism of 80% of commonly used drugs, including
relationship where microbes have access to food components and ha- anticancer molecules, and therefore, CYP450 inhibitors are presently
bitable gut environment while simultaneously providing humans ben- being investigated to improve the pharmacokinetics of several antic-
eficial by-product metabolites. A disbalance in this relationship has ancer drugs [55].
been shown to lead to inflammatory bowel disease and colorectal Langhammer and Nilsen [56] found that ginger extracts obtained
cancer. Diet therefore plays an essential role in establishing overall from commercially available ginger capsule inhibited CYP activities
health. such as CYP1A2, CYP2D6, CYP3A4, with IC50 scores of 320, 445, and
The various ways ginger can be employed to prevent/manage/ and/ 565 μg/mL, respectively. In addition, it was [57] observed that 6-, 8-,
or treat gastrointestinal cancer has been reviewed in many occasions and 10-gingerol blocked the action of CYP450 activity. The three gin-
[36–40]. Most recently, Ganaie et al [41] investigated the chemo-pre- gerols exhibited potent inhibition on CYP2C9 activity, modest inhibi-
ventive efficacy of zingerone on colon carcinogenesis in Wistar rats. The tion on CYP2C19 and CYP3A4, and weakly inhibited CYP2D6. Among
four groups of rodents were given normal saline (control), 1,2-di- them, 8-gingerol showed the highest inhibition of P450 enzymes dis-
methylhydrazine (dose rate used was 20 mg/kg), zingerone (dose rate playing IC50 values of 6.8, 12.5, 8.7, and 42.7 μmol/L against CYP2C9,
20 mg/kg) and zingerone again (dose rate 100 mg/kg). Key findings CYP2C19, CYP3A4, and CYP2D6, respectively. Another study by
were the decreased activity of cytochrome P4502E1 and decreased ROS Mukkavilli et al [58] also found that 6-, 8-, and 10-gingerol, and 6-
level with the ingestion of zingerone meaning that supplementation of shogaol repressed the main membrane-bound players of the CYP450
zingerone has the capability to be used as a chemopreventive agent. enzyme system, including CYP1A2, CYP2A6, CYP2B6, CYP2C8,
Ganaie et al [41] also discussed the potential of zingerone to regulate CYP2C9, CYP2C19, CYP2D6, and CYP3A enzymes.
action of xenobiotic enzymes of Phase I cytochrome P4502E1 and its
ability to suppressed NF-kB-p65, COX-2, iNOS and PCNA, Ki-67. 4.2. Cyclooxygenase (COX)
Similarly, Wang et al [42] found that the gut microbiome has an
essential function in the metabolic pathway of 6-shogaol by reducing Cyclooxygenase enzymes (specifically COX-2) are key molecular
the double bond and ketone group. They showed that 6-shogaol and its targets for cancer intervention at both early and late stages. COX-2 is
metabolites interacts with the human gut microbiota and in mice mi- over-expressed during carcinogenesis, and seems to be involved in the
crobiota through various chemical reactions in the intestines. As a re- start and progression of tumor [59]. 10- gingerol, 8-shogaol, and 10-
sult, the products of the chemicals processes adds to the anti-in- shogaol were found to hinder COX-2 with IC50 scores of 32, 17.5 and
flammation characteristics of ginger which are key in its anti-cancerous 7.5 μM, respectively [60]. In another study by Tjendraputra et al [61],
activities with many studies linking ginger to inhibition of free radical they observed that 10-gingerol, 6-shogaol, and 8-paradol exhibited

5
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

potent COX-2 inhibition with IC50 values of 3.7, 2.1, 3.4 μM, respec- 4.5. Leukotriene A4 hydrolase
tively. In addition, 8-gingerdiol (IC50 = 12.5 μM), 8- gingerol
(IC50 = 10 μM), and its positional isomer (IC50 = 15.8 μM) displayed The proinflammatory enzyme, leukotriene A4 hydrolase (LTA4H), is
comparatively strong COX-2 inhibitory activity. involved in the biosynthesis of leukotriene B4 (LTB4), a chemo-at-
tractant which causes an intense inflammatory reaction associated with
4.3. Matrix metalloproteinases cancer development. LTA4H is reported to be highly expressed in col-
orectal carcinoma.
Matrix metalloproteinases (MMPs) is a family of structurally related El-Naggar et al [68] found that methyl shogaol and 4′-O-prenyl- [6]-
zinc dependent endopeptidases which are key players for extracellular gingerol displayed high LTA4H aminopeptidase (IC50 = 4.92 and
matrix degradation, hence, are associated with the invasion and me- 3.01 μM, respectively) and epoxide hydrolase inhibitory activities
tastasis of tumor. So far, more than 20 human MMPs have been ac- (IC50 = 11.27 & 7.25 μM, respectively). Among the tested gingerols, 10-
knowledged, among which, MMP-2 and -9 are thought to be most sig- gingerol was the most efficient inhibitor of LTA4H aminopeptidase
nificant in tumor invasion due to their role in the degradation of type IV (IC50 = 21.59 μM) and epoxide hydrolase (IC50 = 15.24 μM). These
collagen. Currently, agents reducing the expression of either MMP-2 or results corroborates with the observed in vitro cytotoxic effect of ginger
MMP-9 have been found to effectively inhibit cancer cell invasion [62]. and isolated bioactive phytochemicals against colorectal cancer cells, as
Interestingly, 6-shogaol was found to dose-dependently decrease MMP- mentioned above, which indicates their potential as natural inhibitors
9 gene activation, protein expression and secretion by blocking nuclear of aminopeptidase and epoxide hydrolase for colorectal cancer.
factor-kB activation [62]. Zingerone also displayed strong anti-angio-
genic activity by inhibiting MMP-2 and MMP-9 during tumor progres- 5. Combinational therapy
sion [63].
Moreover, Weng et al [64] observed that treatment of HepG2 and The combination of two or more types of treatment to specifically
Hep3B cells with 6-shogaol (1, 2.5, 5, and 10 μM) or 6-gingerol (5, 10, target cancer-inducing or cell sustaining pathways has become a fun-
25, and 50 μM), respectively, suppressed MMP-9 activity in a dose-de- damental of cancer therapy. This approach can potentially act in a sy-
pendent manner while the activity of MMP-2 was not significantly nergistic and/or additive way and decreases drug resistance and toxi-
changed. Upon treatment of HepG2 cells for 24 h with 0–10 μM 6- city [69].
shogaol or 0–50 μM 6-gingerol, the mRNA expression of MMP9 was In the study of Brahmbhatt et al [70], numerous binary blends of
nearly unchanged, except for 1 μM 6-shogaol treatment which caused a ginger biophenolics were tested at several concentrations against the
dramatic reduction in the levels of MMP-9 mRNA. On the other hand, human prostate cancer PC-3 cells. They found that cell proliferation
24 h treatment of 6-shogaol or 6-gingerol in Hep3B cells reduced the could be hindered by binary combinations of ginger bioactives. 83% of
mRNA expression of MMP-9. Nonetheless, a decrease in MMP-9 mRNA gingerol combinations (6-gingerol+8-gingerol, 8-gingerol+10-gin-
level was only observed after treatment with 6-gingerol. gerol, 6-gingerol+10-gingerol) inhibited PC-3 proliferation in a sy-
Additionally, it was found that 6-gingerol treatment reduced MMP-2 nergistic manner. Another example is the grouping gingerol (90%) and
and MMP-9 activities in MDA-MB-231 cells [65]. Also, 6-gingerol, at 6-shogaol (6-shogaol+6-gingerol, 6-shogaol+8-gingerol, and 6-sho-
levels above 5 μM, was able to reduce the quantity of MMP-2 protein in gaol+10-gingerol) displayed synergism. Very strong synergism was
the culture supernatant, but the level of MMP-9 protein remained un- exhibited by 80% of the 6-gingerol+6-shogaol combinations. Due to
changed. However, 6-gingerol reduced the mRNA expression of both the ginger matrix complexity and minute amount of biophenolics, the
MMP-2 and MMP-9. researchers further investigated if increasing their individual amounts
in the ginger extract could cause an enhanced antiproliferative effect.
4.4. Telemerase They observed that, specifically, ginger extract+6-gingerol and ginger
extract+10-gingerol in combination displayed remarkable synergistic
Telomerase is another key target in cancer therapy. Without telo- antiproliferative activity.
merase, the length of telomeres of typical somatic cells are reduced at Furthermore, Hakim et al [71] showed that ginger extract (aqueous)
every cell division. When a telomere is shortened to a critical length at causes a greater inhibitory effect on the division of HCT 116 colorectal
the cellular level, that cell is prompted to cellular senescence. cancer cells (IC50 = 3 mg/mL) as opposed to Gelam honey
Therefore, cellular senescence induced by telomere reduction is re- (IC50 = 75 mg/mL). Interestingly, the collective action of the two nat-
garded as a tumor suppressor mechanism. However, more than 85% of ural agents (3 mg/mL ginger+75 mg/mL Gelam honey) synergistically
all cancers are able of maintaining the length of their telomeres by reduced the IC50 of Gelam honey (22 mg/mL). Extract of ginger was
reactivating telomerase to prevent cancer cells from the replicative also found to enhance the apoptosis effect of 5-fluorouracil.
senescence [66,67]. Additionally, combination of γ-Tocotrienol and 6-gingerol sy-
Navakoon et al [66] showed that long-term incubation with sub- nergistically induced cytotoxicity and apoptosis in HT-29 and SW837
cytotoxic concentration of ginger extract (ethyl acetate fraction), which colorectal cancer cells, corresponding to IC50 values of 105 and 70 μg/
allowed A549 cells to multiply normally, stimulated telomere reduction mL, respectively. Additional benefits were a rise in apoptosis after 24 h
which subsequently induced cellular senescence in these cells, de- of treatment (21.2% and 55.4% in HT-29 and SW837, respectively)
monstrated by an upsurge in the senescence associated β‑galactosidase with unaffected normal hepatic cells and morphological changes such
positive cells and a decrease in clonogenicity. as cell shrinkage and pyknosis [72]. Combination of 6-gingerol (45 μM)
The transcriptional regulation of telomerase reverse transcriptase in with paclitaxel (0.36 μM) was able to reduce 83.2% growth of HeLa
human (hTERT) also tend to be a major mechanism towards controlling cells, which was more effective compared to 6- gingerol (45 μM) with 5-
the activity of telomerase. c-Myc is one transcription factor, which, col- FU (22.5 μM) (52% inhibition) or 6-gingerol treatment alone (10.75%
lectively with Max, binds to the E-box on the hTERT promoter, thereby inhibition) [23]. In addition, combined treatment of zingerone and its
activating hTERT expression [67]. Tuntiwechapikul et al [67] observed novel derivative synergistically suppressed hepatocellular carcinoma
that ginger extract (ethyl acetate) can time- and concentration-depen- metastasis by the inhibition of the TGF-β1 induced epithelial-me-
dently inhibit the activity of the two main molecular targets of cancer, senchymal transition and suppressed migration and invasion of hepa-
hTERT and c-Myc, in A549 lung cancer cells. The cancer cells displayed tocellular [73].
reduced telomerase activity due to decreased protein manufacture rather Rahman et al [74] studied the anticancer properties of a number of
than direct down regulation of telomerase. The decrease of hTERT ex- compounds including 6-gingerol, epigallocatechin gallate (EGCG),
pression tends to concur with the decrease of c-Myc activity. asiaticoside (AS) and vitamin E (tocotrienol-rich fraction (TRF)). EGCG

6
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

+ 6-gingerol synergistically induced apoptosis and inhibited the blood circulation system and enough time to reach the target tumor
growth of 1321N1 and LN18 glioma cancer cells. On the other hand, all cell. Similarly, Behroozeh et al [103] observed that PEGylated nano-
tested combinations (TRF + 6-gingerol, TRF + EGCG and EGCG + 6- niosomal gingerol showed higher cytotoxicity on T47D breast cancer
gingerol) were reported to be antagonistic on SW1783. Moreover, Ko- cell than the standard drug. More than 76% of the gingerol drug were
towski et al [75] found that although 6‐shogaol combined with cisplatin enclosed in the PEGylated nanoniosomal formulation and the gingerol
displayed an antagonistic effect, when coupled with irradiation ex- release from the nanoparticles consisted of an instant and fast release
hibited a synergistic decrease of clonogenic survival in head and neck phase at the start and then a slower release phase. Applications have
squamous cell carcinoma cell lines. also been carried out in the treatment of lung cancer [104].
Moreover, gingerol synergised cytotoxicity of doxorubicin on liver 6-shogaol enriched liposomes in a DMPG-Na carrier also showed
cancer cells without influencing the cellular pharmacokinetics. Also, enhanced in vitro and in vivo anticancer action. The release of 6-shogaol
gingerol decreased the IC50 of doxorubicin against HepG2 and Huh7 from the liposomes was slower than that from ginger oleoresin at all the
liver by 10- and 4-fold, respectively. On top of that, treatment with only tested pH ranges, which showed reduced outcome on the dissolution of
doxorubicin induced cell growth at S-phase and G2/M-phase, while its 6-shogaol from the inner core compartment of the liposomes. In addi-
blending with gingerol arrested cell cycle at the G2/M-phase. In addi- tion, the 6-shogaol rich ginger oleoresin loaded liposomes showed
tion, co-incubation with 6-gingerol (30 μM) caused a complete blockage better cytotoxic effect than the 6-shogaol rich ginger oleoresin and the
of the exaggerated vasoconstriction and impaired vascular relaxation blank liposomes, which can be justified to a greater cellular uptake of
caused by doxorubicin [76]. In addition, ElAshmawy et al [77] reported liposomes through phagocytosis or the fusion process of lipid lipo-
that co-treatment of ginger extract (100 mg/kg orally day after day) somes. Moreover, the 6-shogaol rich ginger oleoresin loaded liposomes,
alongside doxorubicin (4 mg/kg i.p. for 4 cycles every 5 days), ad- (100, 200 and 400 mg/kg body weight) caused an increase in life span
ministered to mice starting on the 12th day of inoculation of Ehrlich in mice with Dalton’s Ascitic Lymphoma up to 91.5%, 92.5% and
ascites carcinoma cells, markedly amplified survival rate, reduced 93.5%, respectively, as opposed to the batch challenged with only the
tumor volume, raised the level of phosphorylated AMPK (PAMPK), and 6-shogaol rich ginger oleoresin (78%, 81% and 84%, respectively)
improved related pathways in mice compared to doxorubicin group. [105].
Moreover, histopathological findings showed improved apoptosis and Additionally, Zhang et al [106] showed the specificity of nano-
absence of multinucleated cells in tumor tissue of the ginger ex- particles, functionalized with ginger bioactives, to colon cancer cells
tract + doxorubicin group. and the stability and non-toxicity of the ingested nanoparticles in so-
A number of clinical trials also proved that supplementation of lutions similar to the stomach and intestine matrices and conditions.
ginger helps to decrease the severity of acute chemotherapy-induced Mouse colitis models showed that the nanoparticles were mainly ab-
nausea in cancer patients [78–81]. Nausea and vomiting are among the sorbed in the intestinal epithelial cells and macrophages and were ef-
most common and troubling adverse effects associated with che- fective in preventing colitis associated cancer. Both tumor numbers per
motherapy [79]. Nevertheless, one of the likely side effect of radio- mouse and tumor loads were significantly decreased by the ginger de-
therapy to the neck and head area is xerostomia, which is a subjective rived nanoparticles treatment of azoxymethane/dextran sodium sulfate
feeling of dry mouth accompanied by decreased salivation. Shooriabi colitis-associated cancer-induced mice. In addition, the level of pro-
et al [82] observed that ginger can improve the xerostomia symptoms inflammatory cytokine and cyclin D1 mRNA were also decreased,
by increasing the rate of salivary secretion in patients undergoing suggesting that a reduction in cell proliferation during colitis-associated
radiotherapy. Nevertheless, Chamani et al [83], reported that ginger cancer development.
decreased the severity of dry mouth in patients with post-radiation Zhang et al [107] further reconstructed the lipid groups of ginger
xerostomia but did not improve dry mouth symptoms or the patients’ functionalized nanoparticles to ginger-derived nanovectors (GDNVs) to
quality of life. behave as a delivery platform for doxorubicin in the treatment of colon
cancer. Comparable to the previously functionalized ginger nano-
6. Nanotherapeutic application particles, the GDNVs were proven to be non-toxic and absorbed by
cancer cells in the large intestines and more importantly the GDNVs
The emerging field of nanomedicine has the ability to change the have the capacity to encapsulate doxorubicin (95.9% at a ginger lipid
way cancer therapy is being provided [84,85]. By targeting the vascular concentration of 100 μmol/L), and displayed a better pH-dependent
tissue and cellular characteristics exclusive to cancer cells [86], nano- drug-release profile compared to commercially available liposomal-
medicine ensures that the drugs are delivered at the right sites, there- doxorubicin. On top of that, the doxorubicin-GDNVs were very stable at
fore making drug delivery and absorption more successful [87,88]. 4 °C (up to 25 days), retained their ability to load doxorubicin, and
Even though the potential of nanotechnology in the treatment/man- exhibited a time-dependent release profile which decreased the release
agement of cancer is a recent and novel development [89,90], sig- of doxorubicin from GDNVs and resulted to a sustained cytotoxic effect
nificant research has been done in the area in order to optimize its against colon cancer cells.
mechanism [91], pharmacokinetics [92] and delivery [93] as well as The researchers then generated nanoparticles with a high affinity to
the searches for new nano-composites such as graphene [94,95] or Colon-26 tumors in vivo by modifying the ginger-derived nanoparticles
hybrid polymer-metal composites for drug delivery [96]. Nanomedicine to incorporate folic acid, a molecule showing high affinity for folate
reduces the chances of developing cancer cells that are resistant [97], receptors. These receptors are highly expressed on many types of tu-
and more importantly, it may allow for more than one type of cancer to mors, while being almost undetectable on healthy cells. The authors
be targeted during one treatment [98]. demonstrated that the folic acid nanovectors efficiently loaded and
The benefits and therapeutic applications of liposomal based drug delivered doxorubicin into cancer cells and inhibited tumor growth.
delivery have been reviewed [99,100] and while a vast number of ap-
plications are in colon cancer, its significance in oral cancer therapy is 7. Conclusion
also discussed [101]. In particular, the work carried out by Khalili et al
[102] found that PEGylated nanoliposomal formulation of gingerol The current review showcases the cytotoxicity propensities of ginger
increased its cytotoxic effect against breast cancer MCF-7 cell at a re- and its main bioactive compounds to a number of cancer types with
duced concentration, allowing a slower drug release, when compared particular attention on breast, cervical, colorectal, leukemia, liver, lung,
with liposomal and standard gingerol. PEGylation allowed a controlled nasopharyngeal, ovarian, prostate, and retinoblastoma types. Further
drug release and protect the liposome against the immune system and evidence is presented to show that apoptotic mechanisms are depen-
lipase enzymes which cause lipid solvation, thereby enabling a longer dent on cancer type. Ginger and its active compounds also exerted

7
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

inhibitory effect against key enzymes linked to cancer progression in- D.P. Mishra, A.A. Mahdi, Anticancer and antioxidant activity of Zingiber officinale
cluding cytochrome 450, cyclooxygenase-2, matrix metalloproteinase-2 Roscoe rhizome, Indian J. Exp. Biol. 54 (2016) 767–773.
[18] B.S. Tan, O. Kang, C.W. Mai, K.H. Tiong, A.S.-B. Khoo, M.R. Pichika,
and -9, telomerase, and leukotriene A4 hydrolase. In addition, an en- T.D. Bradshaw, C.-O. Leong, 6-Shogaol inhibits breast and colon cancer cell pro-
hancement in the efficacy of chemotherapeutic drugs and reduction of liferation through activation of peroxisomal proliferator activated receptor γ
the side effects of radiotherapy was exhibited by ginger. More inter- (PPARγ), Cancer Lett. 336 (1) (2013) 127–139.
[19] A.C.B. Martin, A.M. Fuzer, A.B. Becceneri, J.A. da Silva, R. Tomasin, D. Denoyer,
estingly however, future work remains to be done to provide a com- S.-H. Kim, K.A. McIntyre, H.B. Pearson, B. Yeo, [10]-gingerol induces apoptosis
prehensive understanding of the interactions of the bioactive phyto- and inhibits metastatic dissemination of triple negative breast cancer in vivo,
chemicals in ginger in the human body system through in-vivo Oncotarget 8 (42) (2017) 72260.
[20] Q. Liu, Y.-B. Peng, L.-W. Qi, X.-L. Cheng, X.-J. Xu, L.-L. Liu, E.-H. Liu, P. Li, The
preclinical trials. Also, the application of nanotechnology seems to be a cytotoxicity mechanism of 6-Shogaol-treated HeLa human cervical cancer cells
better delivery system for ginger compounds. Nonetheless, further revealed by label-free shotgun proteomics and bioinformatics analysis, Evid. Based
pharmacodynamic and pharmacokinetic studies to determine the mo- Complement. Altern. Med. 2012 (2012).
[21] Y. Lee, Cytotoxicity evaluation of essential oil and its component from Zingiber
lecular pathway exhibited by these nano-formulations and their bioa-
officinale Roscoe, Toxicol. Res. 32 (3) (2016) 225–230.
vailability are needed. Last but not least, since ginger is traditionally [22] F. Peng, Q. Tao, X. Wu, H. Dou, S. Spencer, C. Mang, L. Xu, L. Sun, Y. Zhao, H. Li,
used against cancer, there is a need for more human clinical trials to Cytotoxic, cytoprotective and antioxidant effects of isolated phenolic compounds
validate the exact dose and mode of administration as practiced tradi- from fresh ginger, Fitoterapia 83 (3) (2012) 568–585.
[23] F. Zhang, J.-G. Zhang, J. Qu, Q. Zhang, C. Prasad, Z.-J. Wei, Assessment of anti-
tionally to establish its efficacy and observe any possible adverse re- cancerous potential of 6-gingerol (Tongling White Ginger) and its synergy with
actions. drugs on human cervical adenocarcinoma cells, Food Chem. Toxicol. 109 (2017)
910–922.
[24] A. Saha, J. Blando, E. Silver, L. Beltran, J. Sessler, J. DiGiovanni, 6-Shogaol from
Funding support dried ginger inhibits growth of prostate cancer cells both in vitro and in vivo
through inhibition of STAT3 and NF-κB signaling, Cancer Prev. Res. 7 (6) (2014)
No specific funding was disclosed. 627–638.
[25] H.-W. Kim, D.-H. Oh, C. Jung, D.-D. Kwon, Y.-C. Lim, Apoptotic effects of 6-gin-
gerol in LNCaP human prostate cancer cells, Soonchunhyang Med. Sci. 17 (2)
Declaration of Competing Interest (2011) 75–79.
[26] L. Lv, H. Chen, D. Soroka, X. Chen, T. Leung, S. Sang, 6-Gingerdiols as the major
metabolites of 6-gingerol in cancer cells and in mice and their cytotoxic effects on
The authors made no disclosures. human cancer cells, J. Agric. Food Chem. 60 (45) (2012) 11372–11377.
[27] J.-Y. Hung, Y.-L. Hsu, C.-T. Li, Y.-C. Ko, W.-C. Ni, M.-S. Huang, P.-L. Kuo, 6-
References Shogaol, an active constituent of dietary ginger, induces autophagy by inhibiting
the AKT/mTOR pathway in human non-small cell lung cancer A549 cells, J. Agric.
Food Chem. 57 (20) (2009) 9809–9816.
[1] L.-Q. Peng, J. Cao, L.-J. Du, Q.-D. Zhang, J.-J. Xu, Y.-B. Chen, Y.-T. Shi, R.-R. Li, [28] S.H.M. Habib, S. Makpol, N.A.A. Hamid, S. Das, W.Z.W. Ngah, Y.A.M. Yusof,
Rapid ultrasonic and microwave-assisted micellar extraction of zingiberone, sho- Ginger extract (Zingiber officinale) has anti-cancer and anti-inflammatory effects
gaol and gingerols from gingers using biosurfactants, J. Chromatogr. A 1515 on ethionine-induced hepatoma rats, Clinics 63 (6) (2008) 807–813.
(2017) 37–44. [29] P. Santos, G. Avanço, S. Nerilo, R. Marcelino, V. Janeiro, M. Valadares,
[2] J. Prakash, Chemical composition and antioxidant properties of ginger root M. Machinski, Assessment of cytotoxic activity of rosemary (Rosmarinus officinalis
(Zingiber officinale), J. Med. Plants Res. 4 (24) (2010) 2674–2679. L.), turmeric (Curcuma longa L.), and ginger (Zingiber officinale R.) essential oils
[3] D. Pan, C. Zeng, W. Zhang, T. Li, Z. Qin, X. Yao, Y. Dai, Z. Yao, Y. Yu, X. Yao, Non- in cervical cancer cells (HeLa), Sci. World J. 2016 (2016).
volatile pungent compounds isolated from Zingiber officinale and their mechan- [30] Q. Liu, Y.-B. Peng, P. Zhou, L.-W. Qi, M. Zhang, N. Gao, E.-H. Liu, P. Li, 6-Shogaol
isms of action, Food Funct. 10 (2) (2019) 1203–1211. induces apoptosis in human leukemia cells through a process involving caspase-
[4] B.H. Ali, G. Blunden, M.O. Tanira, A. Nemmar, Some phytochemical, pharmaco- mediated cleavage of eIF2α, Mol. Cancer 12 (1) (2013) 135.
logical and toxicological properties of ginger (Zingiber officinale Roscoe): a review [31] S.H. Lee, M. Cekanova, S.J. Baek, Multiple mechanisms are involved in 6‐gin-
of recent research, Food Chem. Toxicol. 46 (2) (2008) 409–420. gerol‐induced cell growth arrest and apoptosis in human colorectal cancer cells,
[5] Z. Li, Y. Wang, M. Gao, W. Cui, M. Zeng, Y. Cheng, J. Li, Nine new gingerols from Mol. Carcinog. 47 (3) (2008) 197–208.
the rhizoma of Zingiber officinale and their cytotoxic activities, Molecules 23 (2) [32] E. Radhakrishnan, S.V. Bava, S.S. Narayanan, L.R. Nath, A.K.T. Thulasidasan,
(2018) 315. E.V. Soniya, R.J. Anto, [6]-Gingerol induces caspase-dependent apoptosis and
[6] Y. Liu, R.J. Whelan, B.R. Pattnaik, K. Ludwig, E. Subudhi, H. Rowland, prevents PMA-induced proliferation in colon cancer cells by inhibiting MAPK/AP-
N. Claussen, N. Zucker, S. Uppal, D.M. Kushner, Terpenoids from Zingiber offici- 1 signaling, PLoS One 9 (8) (2014) e104401.
nale (Ginger) induce apoptosis in endometrial cancer cells through the activation [33] L.-W. Qi, Z. Zhang, C.-F. Zhang, S. Anderson, Q. Liu, C.-S. Yuan, C.-Z. Wang, Anti-
of p53, PLoS One 7 (12) (2012) e53178. colon cancer effects of 6-shogaol through G2/M cell cycle arrest by p53/p21-cdc2/
[7] M.-J. Ko, H.-H. Nam, M.-S. Chung, Conversion of 6-gingerol to 6-shogaol in ginger cdc25A crosstalk, Am. J. Chin. Med. 43 (04) (2015) 743–756.
(Zingiber officinale) pulp and peel during subcritical water extraction, Food Chem. [34] Y. Zhu, R.F. Warin, D.N. Soroka, H. Chen, S. Sang, Metabolites of ginger compo-
270 (2019) 149–155. nent [6]-shogaol remain bioactive in cancer cells and have low toxicity in normal
[8] R.B. Semwal, D.K. Semwal, S. Combrinck, A.M. Viljoen, Gingerols and shogaols: cells: chemical synthesis and biological evaluation, PLoS One 8 (1) (2013) e54677.
important nutraceutical principles from ginger, Phytochemistry 117 (2015) [35] L. Gentschew, L.R. Ferguson, Role of nutrition and microbiota in susceptibility to
554–568. inflammatory bowel diseases, Mol. Nutr. Food Res. 56 (4) (2012) 524–535.
[9] J.-P. Ma, X.-L. Jin, L. Yang, Z.-L. Liu, Two new diarylheptanoids from the rhizomes [36] M. Levy, E. Blacher, E. Elinav, Microbiome, metabolites and host immunity, Curr.
of Zingiber officinale, Chin. Chem. Lett. 15 (11) (2004) 1306–1308. Opin. Microbiol. 35 (2017) 8–15.
[10] R. Silambarasan, M. Ayyanar, An ethnobotanical study of medicinal plants in [37] Y.K. Lee, S.K. Mazmanian, Has the microbiota played a critical role in the evo-
Palamalai region of Eastern Ghats, India, J. Ethnopharmacol. 172 (2015) 162–178. lution of the adaptive immune system? Science 330 (6012) (2010) 1768–1773.
[11] R. Silambarasan, J. Sureshkumar, J. Krupa, S. Amalraj, M. Ayyanar, Traditional [38] K. Singletary, Ginger: an overview of health benefits, Nutr. Today 45 (4) (2010)
herbal medicines practiced by the ethnic people in Sathyamangalam forests of 171–183.
Western Ghats, India, Eur. J. Integr. Med. 16 (2017) 61–72. [39] P.J. Turnbaugh, R.E. Ley, M. Hamady, C.M. Fraser-Liggett, R. Knight, J.I. Gordon,
[12] Y.-Y. Siew, S. Zareisedehizadeh, W.-G. Seetoh, S.-Y. Neo, C.-H. Tan, H.-L. Koh, The human microbiome project, Nature 449 (7164) (2007) 804.
Ethnobotanical survey of usage of fresh medicinal plants in Singapore, J. [40] S. Prasad, A.K. Tyagi, Ginger and its constituents: role in prevention and treatment
Ethnopharmacol. 155 (3) (2014) 1450–1466. of gastrointestinal cancer, Gastroenterol. Res. Pract. 2015 (2015) 11.
[13] N.A. Jaradat, R. Shawahna, A.M. Eid, R. Al-Ramahi, M.K. Asma, A.N. Zaid, Herbal [41] M.A. Ganaie, A. Al Saeedan, H. Madhkali, B.L. Jan, T. Khatlani, I.A. Sheikh,
remedies use by breast cancer patients in the West Bank of Palestine, J. M.U. Rehman, K. Wani, Chemopreventive efficacy zingerone (4‐[4‐hydro-
Ethnopharmacol. 178 (2016) 1–8. xy‐3‐methylphenyl] butan‐2‐one) in experimental colon carcinogenesis in Wistar
[14] M.S. Ali-Shtayeh, R.M. Jamous, N.M. Salameh, R.M. Jamous, A.M. Hamadeh, rats, Environ. Toxicol. 34 (2019) 610–625.
Complementary and alternative medicine use among cancer patients in Palestine [42] C.-Z. Wang, L.-W. Qi, C.-S. Yuan, Cancer chemoprevention effects of ginger and its
with special reference to safety-related concerns, J. Ethnopharmacol. 187 (2016) active constituents: potential for new drug discovery, Am. J. Chin. Med. 43 (07)
104–122. (2015) 1351–1363.
[15] F. Kabbaj, B. Meddah, Y. Cherrah, E. Faouzi, Ethnopharmacological profile of [43] G.H. Park, J.H. Park, H.M. Song, H.J. Eo, M.K. Kim, J.W. Lee, M.H. Lee, K.-H. Cho,
traditional plants used in Morocco by cancer patients as herbal therapeutics, J.R. Lee, H.J. Cho, Anti-cancer activity of Ginger (Zingiber officinale) leaf through
Phytopharmacology 2 (2) (2012) 243–256. the expression of activating transcription factor 3 in human colorectal cancer cells,
[16] C. Agyare, V. Spiegler, A. Asase, M. Scholz, G. Hempel, A. Hensel, An ethno- BMC Complement. Altern. Med. 14 (1) (2014) 408.
pharmacological survey of medicinal plants traditionally used for cancer treat- [44] I. Karaboz, Antimicrobial and cytotoxic activities of Zingiber officinalis extracts,
ment in the Ashanti region, ghana, J. Ethnopharmacol. 212 (2018) 137–152. FABAD J. Pharm. Sci 33 (2010) 76–85.
[17] J.A. Ansari, M.K. Ahmad, A.R. Khan, N. Fatima, H.J. Khan, N. Rastogi, [45] M. Kumara, M. Shylajab, P. Nazeemc, T. Babu, 6-Gingerol is the most potent

8
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

anticancerous compound in ginger (Zingiber officinale Rosc.), J. Dev. Drugs 6 10280–10297.


(2017) 6–11. [73] Y.-J. Kim, Y. Jeon, T. Kim, W.-C. Lim, J. Ham, Y.N. Park, T.-J. Kim, H. Ko,
[46] S. Abdullah, S.A.Z. Abidin, N.A. Murad, S. Makpol, W.Z.W. Ngah, Y.A.M. Yusof, Combined treatment with zingerone and its novel derivative synergistically in-
Ginger extract (Zingiber officinale) triggers apoptosis and G0/G1 cells arrest in hibits TGF-β1 induced epithelial-mesenchymal transition, migration and invasion
HCT 116 and HT 29 colon cancer cell lines, Afr. J. Biochem. Res. 4 (5) (2010) of human hepatocellular carcinoma cells, Bioorg. Med. Chem. Lett. 27 (4) (2017)
134–142. 1081–1088.
[47] C.-Y. Chen, Y.-W. Li, S.-Y. Kuo, Effect of [10]-gingerol on [ca2+] i and cell death [74] A. Rahman, S. Makpol, R. Jamal, R. Harun, N. Mokhtar, W. Ngah, Tocotrienol-rich
in human colorectal cancer cells, Molecules 14 (3) (2009) 959–969. fraction,[6]-gingerol and epigallocatechin gallate inhibit proliferation and induce
[48] M.A. Al-Tamimi, B. Rastall, I.M. Abu-Reidah, Chemical composition, cytotoxic, apoptosis of glioma cancer cells, Molecules 19 (9) (2014) 14528–14541.
apoptotic and antioxidant activities of main commercial essential oils in Palestine: [75] U. Kotowski, L. Kadletz, S. Schneider, E. Foki, R. Schmid, R. Seemann,
a comparative study, Medicines 3 (4) (2016) 27. D. Thurnher, G. Heiduschka, 6‐shogaol induces apoptosis and enhances radio-
[49] K. Jeena, V.B. Liju, R. Kuttan, Antitumor and cytotoxic activity of ginger essential sensitivity in head and neck squamous cell carcinoma cell lines, Phytother. Res. 32
oil (Zingiber officinale Roscoe), Int. J. Pharm. Pharm. Sci. 7 (8) (2015) 341–344. (2) (2018) 340–347.
[50] R. Pashaei-Asl, F. Pashaei-Asl, P.M. Gharabaghi, K. Khodadadi, M. Ebrahimi, [76] F. Al-Abbasi, E. Alghamdi, M. Baghdadi, A. Alamoudi, A. El-Halawany, H. El-
E. Ebrahimie, M. Pashaiasl, The inhibitory effect of ginger extract on Ovarian Bassossy, A. Aseeri, A. Al-Abd, Gingerol synergizes the cytotoxic effects of dox-
cancer cell line; application of systems biology, Adv. Pharm. Bull. 7 (2) (2017) orubicin against liver cancer cells and protects from its vascular toxicity,
241. Molecules 21 (7) (2016) 886.
[51] H. Rashid, G. Umamaheswari, Evaluation of the cytotoxic effect of ginger extract [77] N.E. El-Ashmawy, N.F. Khedr, H.A. El-Bahrawy, H.E.A. Mansour, Ginger extract
against prostate Cancer model using in vitro, World J. Pharm. Pharm. Sci. 6 (2017) adjuvant to doxorubicin in mammary carcinoma: study of some molecular me-
1044–1053. chanisms, Eur. J. Nutr. 57 (3) (2018) 981–989.
[52] P. Karna, S. Chagani, S.R. Gundala, P.C. Rida, G. Asif, V. Sharma, M.V. Gupta, [78] W. Marx, A.L. McCarthy, K. Ried, L. Vitetta, D. McKavanagh, D. Thomson, A. Sali,
R. Aneja, Benefits of whole ginger extract in prostate cancer, Br. J. Nutr. 107 (4) L. Isenring, Can ginger ameliorate chemotherapy-induced nausea? Protocol of a
(2012) 473–484. randomized double blind, placebo-controlled trial, BMC Complement. Altern.
[53] B. Meng, H. Ii, W. Qu, H. Yuan, Anticancer effects of gingerol in retinoblastoma Med. 14 (1) (2014) 134.
Cancer cells (RB355 cell line) are mediated via apoptosis induction, cell cycle [79] Y. Panahi, A. Saadat, A. Sahebkar, F. Hashemian, M. Taghikhani, E. Abolhasani,
arrest and upregulation of PI3K/Akt signaling pathway, Med. Sci. Monit. 24 Effect of ginger on acute and delayed chemotherapy-induced nausea and vomiting:
(2018) 1980. a pilot, randomized, open-label clinical trial, Integr. Cancer Ther. 11 (3) (2012)
[54] M.H. Baig, M. Adil, R. Khan, S. Dhadi, K. Ahmad, G. Rabbani, T. Bashir, 204–211.
M.A. Imran, F.M. Husain, E.J. Lee, Enzyme Targeting Strategies for Prevention and [80] J.L. Ryan, C.E. Heckler, J.A. Roscoe, S.R. Dakhil, J. Kirshner, P.J. Flynn,
Treatment of Cancer: Implications for Cancer Therapy, Seminars in Cancer J.T. Hickok, G.R. Morrow, Ginger (Zingiber officinale) reduces acute che-
Biology, Elsevier, 2017. motherapy-induced nausea: a URCC CCOP study of 576 patients, Support. Care
[55] B. Mittal, S. Tulsyan, S. Kumar, R.D. Mittal, G. Agarwal, Cytochrome P450 in Cancer 20 (7) (2012) 1479–1489.
Cancer Susceptibility and Treatment, Advances in Clinical Chemistry, Elsevier, [81] F. Sanaati, S. Najafi, Z. Kashaninia, M. Sadeghi, Effect of ginger and chamomile on
2015, pp. 77–139. nausea and vomiting caused by chemotherapy in iranian women with breast
[56] A.J. Langhammer, O.G. Nilsen, In vitro inhibition of human CYP1A2, CYP2D6, and cancer, Asian Pac. J. Cancer Prev. 17 (8) (2016) 4125–4129.
CYP3A4 by six herbs commonly used in pregnancy, Phytother. Res. 28 (4) (2014) [82] M. Shooriabi, D. Ardakani, B. Mansoori, S. Satvati, R. Sharifi, The effect of ginger
603–610. extract on radiotherapy-oriented salivation in patients with xerostomia: a double-
[57] M. Li, P.-z. Chen, Q.-x. Yue, J.-q. Li, R.-a. Chu, W. Zhang, H. Wang, Pungent ginger blind controlled study, Pharm. Lett. 8 (15) (2016) 37–45.
components modulates human cytochrome P450 enzymes in vitro, Acta [83] G. Chamani, M.R. Zarei, M. Mehrabani, N. Nakhaee, B. Kalaghchi, M. Aghili,
Pharmacol. Sin. 34 (9) (2013) 1237. A. Alaee, Assessment of systemic effects of ginger on salivation in patients with
[58] R. Mukkavilli, S.R. Gundala, C. Yang, S. Donthamsetty, G. Cantuaria, G.R. Jadhav, post-radiotherapy xerostomia, J. Oral Health Oral Epidemiol. 6 (3) (2017)
S. Vangala, M.D. Reid, R. Aneja, Modulation of cytochrome P450 metabolism and 130–137.
transport across intestinal epithelial barrier by ginger biophenolics, PLoS One 9 [84] V. Kumar, P.C. Bhatt, M. Rahman, G. Kaithwas, H. Choudhry, F.A. Al-Abbasi,
(9) (2014) e108386. F. Anwar, A. Verma, Fabrication, optimization, and characterization of umbelli-
[59] W.F. Anderson, A. Umar, E.T. Hawk, Cyclooxygenase inhibition in cancer pre- ferone β-D-galactopyranoside-loaded PLGA nanoparticles in treatment of hepato-
vention and treatment, Expert Opin. Pharmacother. 4 (12) (2003) 2193–2204. cellular carcinoma: in vitro and in vivo studies, Int. J. Nanomed. 12 (2017)
[60] R.B. van Breemen, Y. Tao, W. Li, Cyclooxygenase-2 inhibitors in ginger (Zingiber 6747–6758.
officinale), Fitoterapia 82 (1) (2011) 38–43. [85] M. Rahman, M.Z. Ahmad, I. Kazmi, S. Akhter, Y. Kumar, F.J. Ahmad, F. Anwar,
[61] E. Tjendraputra, V.H. Tran, D. Liu-Brennan, B.D. Roufogalis, C.C. Duke, Effect of Novel approach for the treatment of Cancer: theranostic nanomedicine,
ginger constituents and synthetic analogues on cyclooxygenase-2 enzyme in intact Pharmacologia 3 (2012) 371–376.
cells, Bioorg. Chem. 29 (3) (2001) 156–163. [86] A. Sohail, A. Mohammad Zaki, S. Anjali, A. Iqbal, R. Mahfoozur, A. Mohammad,
[62] H. Ling, H. Yang, S.H. Tan, W.K. Chui, E.H. Chew, 6-Shogaol, an active constituent J. Gaurav Kumar, A. Farhan Jalees, K. Roop Krishen, Cancer targeted metallic
of ginger, inhibits breast cancer cell invasion by reducing matrix metalloprotei- nanoparticle: targeting overview, recent advancement and toxicity concern, Curr.
nase-9 expression via blockade of nuclear factor-kappaB activation, Br. J. Pharm. Des. 17 (18) (2011) 1834–1850.
Pharmacol. 161 (8) (2010) 1763–1777. [87] M. Rahman, S. Beg, A. Ahmed, S. Swain, Emergence of functionalized nanome-
[63] W.-Y. Bae, J.-S. Choi, J.-E. Kim, C. Park, J.-W. Jeong, Zingerone suppresses an- dicines in Cancer chemotherapy: recent advancements, current challenges and
giogenesis via inhibition of matrix metalloproteinases during tumor development, toxicity considerations, Recent Pat. Nanomed. 3 (2) (2013) 128–139.
Oncotarget 7 (30) (2016) 47232–47241. [88] A. Preeti, R. Mahfoozur, B. Sarwar, A. Shivali, D. Vishal, C. Rupali, Cancer targeted
[64] C.J. Weng, C.F. Wu, H.W. Huang, C.T. Ho, G.C. Yen, Anti‐invasion effects of magic bullets for effective treatment of Cancer, Recent Pat. Antiinfect. Drug
6‐shogaol and 6‐gingerol, two active components in ginger, on human hepato- Discov. 9 (2) (2014) 121–135.
carcinoma cells, Mol. Nutr. Food Res. 54 (11) (2010) 1618–1627. [89] M.Z. Ahmad, S. Akhter, G.K. Jain, M. Rahman, S.A. Pathan, F.J. Ahmad, R.K. Khar,
[65] H.S. Lee, E.Y. Seo, N.E. Kang, W.K. Kim, [6]-Gingerol inhibits metastasis of MDA- Metallic nanoparticles: technology overview & drug delivery applications in on-
MB-231 human breast cancer cells, J. Nutr. Biochem. 19 (5) (2008) 313–319. cology, Expert Opin. Drug Deliv. 7 (8) (2010) 927–942.
[66] N. Kaewtunjai, W. Pompimon, W. Tuntiwechapikul, Ginger (Zingiber officinale) [90] M. Rahman, M.Z. Ahmad, I. Kazmi, S. Akhter, M. Afzal, G. Gupta, F. Jalees Ahmed,
extract promotes telomere shortening and induces cellular senescence in A549 F. Anwar, Advancement in multifunctional nanoparticles for the effective treat-
lung Cancer cells, Planta Med. Int. Open 4 (S 01) (2017) Mo-PO-12. ment of cancer, Expert Opin. Drug Deliv. 9 (4) (2012) 367–381.
[67] W. Tuntiwechapikul, T. Taka, C. Songsomboon, N. Kaewtunjai, A. Imsumran, [91] M.Z. Ahmad, S. Akhter, M. Anwar, A. Kumar, M. Rahman, A.H. Talasaz,
L. Makonkawkeyoon, W. Pompimon, T.R. Lee, Ginger extract inhibits human F.J. Ahmad, Colorectal cancer targeted Irinotecan-Assam Bora rice starch based
telomerase reverse transcriptase and c-Myc expression in A549 lung cancer cells, microspheres: a mechanistic, pharmacokinetic and biochemical investigation,
J. Med. Food 13 (6) (2010) 1347–1354. Drug Dev. Ind. Pharm. 39 (12) (2013) 1936–1943.
[68] M.H. El-Naggar, A. Mira, F.M.A. Bar, K. Shimizu, M.M. Amer, F.A. Badria, [92] P. Pandey, M. Rahman, P.C. Bhatt, S. Beg, B. Paul, A. Hafeez, F.A. Al-Abbasi,
Synthesis, docking, cytotoxicity, and LTA4H inhibitory activity of new gingerol M.S. Nadeem, O. Baothman, F. Anwar, V. Kumar, Implication of nano-antioxidant
derivatives as potential colorectal cancer therapy, Bioorg. Med. Chem. 25 (3) therapy for treatment of hepatocellular carcinoma using PLGA nanoparticles of
(2017) 1277–1285. rutin, Nanomed. Lond. (Lond) 13 (8) (2018) 849–870.
[69] R.B. Mokhtari, T.S. Homayouni, N. Baluch, E. Morgatskaya, S. Kumar, B. Das, [93] A. Mohammad Zaki, A. Sohail, A. Iqbal, R. Mahfoozur, A. Mohammad,
H. Yeger, Combination therapy in combating cancer, Oncotarget 8 (23) (2017) K.J. Gourav, J.A. Farhan, K. Roop Krishen, Development of polysaccharide based
38022. Colon Targeted drug delivery system: design and evaluation of Assam Bora rice
[70] M. Brahmbhatt, S.R. Gundala, G. Asif, S.A. Shamsi, R. Aneja, Ginger phytochem- starch based matrix tablet, Curr. Drug Deliv. 8 (5) (2011) 575–581.
icals exhibit synergy to inhibit prostate cancer cell proliferation, Nutr. Cancer 65 [94] R. Mahfoozur, A. Mohammad Zaki, A. Javed, F. Jamia, A. Farhan Jalees, M. Gohar,
(2) (2013) 263–272. A.K. Mohammad, A. Sohail, Role of graphene nano-composites in Cancer therapy:
[71] L. Hakim, E. Alias, S. Makpol, W.Z.W. Ngah, N.A. Morad, Y. Yusof, Gelam honey theranostic applications, metabolic fate and toxicity issues, Curr. Drug Metab. 16
and ginger potentiate the anti cancer effect of 5-FU against HCT 116 colorectal (5) (2015) 397–409.
cancer cells, Asian Pac. J. Cancer Prev. 15 (11) (2014) 4651–4657. [95] M. Rahman, S. Akhter, M.Z. Ahmad, J. Ahmad, R.T. Addo, F.J. Ahmad, C. Pichon,
[72] M. Yusof, S. Makpol, R. Jamal, R. Harun, N. Mokhtar, W. Wan Ngah, γ-Tocotrienol Emerging advances in cancer nanotheranostics with graphene nanocomposites:
and 6-Gingerol in combination synergistically induce cytotoxicity and apoptosis in opportunities and challenges, Nanomedicine (Lond) 10 (15) (2015) 2405–2422.
HT-29 and SW837 human colorectal cancer cells, Molecules 20 (6) (2015) [96] S. Beg, M. Rahman, A. Jain, S. Saini, P. Midoux, C. Pichon, F.J. Ahmad, S. Akhter,

9
M.F. Mahomoodally, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Nanoporous metal organic frameworks as hybrid polymer–metal composites for [102] M. Khalili, A. Akbarzadeh, M. Chiani, S. Torabi, The effect of nanoliposomal and
drug delivery and biomedical applications, Drug Discov. Today 22 (4) (2017) PE gylated nanoliposomal forms of 6-gingerol on breast cancer cells, Res. J. Recent
625–637. Sci. 2 (2013) 29–33.
[97] R. Mahfoozur, A. Mohammad Zaki, K. Imran, A. Sohail, A. Muhammad, G. Gaurav, [103] A. Behroozeh, M.M. Tabrizi, S.M. Kazemi, E. Choupani, N. Kabiri, D. Ilbeigi,
S. Vivek Ranjan, Emergence of nanomedicine as Cancer Targeted magic bullets: A.H. Nasab, A.A. Khiyavi, A.S. Kurdi, Evaluation the anti-cancer effect of
recent development and need to address the toxicity apprehension, Curr. Drug PEGylated nano-niosomal gingerol, on breast cancer cell lines (T47D), in-Vitro,
Discov. Technol. 9 (4) (2012) 319–329. Asian Pac. J. Cancer Prevent. APJCP 19 (3) (2018) 645.
[98] J. Kydd, R. Jadia, P. Velpurisiva, A. Gad, S. Paliwal, P. Rai, Targeting strategies for [104] J. Ahmad, S. Akhter, M. Rizwanullah, S. Amin, M. Rahman, M.Z. Ahmad,
the combination treatment of cancer using drug delivery systems, Pharmaceutics 9 M.A. Rizvi, M.A. Kamal, F.J. Ahmad, Nanotechnology-based inhalation treatments
(4) (2017) 46. for lung cancer: state of the art, Nanotechnol. Sci. Appl. 8 (2015) 55–66.
[99] Mahfoozur Rahman, B. Sarwar, V. Amita, K. Imran, P. Dinesh Kumar, A. Firoz, [105] K. Kemkar, S. Lohidasan, A. Sathiyanarayanan, K. Mahadik, 6-shogaol from ginger
A.A.A. Fahad, Vikas Kumar, Therapeutic applications of liposomal based drug oleoresin loaded liposomes using DMPG-Na as a carrier enhances the in-vitro and
delivery and drug targeting for immune linked inflammatory maladies: a con- in-vivo anticancer activity, J. Appl. Pharm. Sci. 8 (2018) 1–10.
temporary view Point, Curr. Drug Targets 18 (13) (2017) 1558–1571. [106] M. Zhang, E. Viennois, M. Prasad, Y. Zhang, L. Wang, Z. Zhang, M.K. Han, B. Xiao,
[100] M. Rahman, V. Kumar, S. Beg, G. Sharma, O.P. Katare, F. Anwar, Emergence of C. Xu, S. Srinivasan, Edible ginger-derived nanoparticles: a novel therapeutic
liposome as targeted magic bullet for inflammatory disorders: current state of the approach for the prevention and treatment of inflammatory bowel disease and
art, Artificial Cells, Nanomed. Biotechnol. 44 (7) (2016) 1597–1608. colitis-associated cancer, Biomaterials 101 (2016) 321–340.
[101] A. Javed, A. Saima, R. Mahfoozur, R. Rehan Abdur, S. Madhur, A. Mohammad [107] M. Zhang, B. Xiao, H. Wang, M.K. Han, Z. Zhang, E. Viennois, C. Xu, D. Merlin,
Zaki, R. Ziyaur, T.A. Richard, A. Farhan Jalees, M. Gohar, K. Mohammad Amjad, Edible ginger-derived nano-lipids loaded with doxorubicin as a novel drug-de-
A. Sohail, Solid matrix based lipidic nanoparticles in oral Cancer chemotherapy: livery approach for colon cancer therapy, Mol. Ther. 24 (10) (2016) 1783–1796.
applications and pharmacokinetics, Curr. Drug Metab. 16 (8) (2015) 633–644.

10

You might also like